WO2015074528A1 - 抗her2抗体及其缀合物 - Google Patents

抗her2抗体及其缀合物 Download PDF

Info

Publication number
WO2015074528A1
WO2015074528A1 PCT/CN2014/091332 CN2014091332W WO2015074528A1 WO 2015074528 A1 WO2015074528 A1 WO 2015074528A1 CN 2014091332 W CN2014091332 W CN 2014091332W WO 2015074528 A1 WO2015074528 A1 WO 2015074528A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
her2
functional fragment
conjugate
cancer
Prior art date
Application number
PCT/CN2014/091332
Other languages
English (en)
French (fr)
Chinese (zh)
Inventor
房健民
黄长江
姜静
姚雪静
Original Assignee
烟台荣昌生物工程有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to KR1020167012018A priority Critical patent/KR101854443B1/ko
Priority to EP18204950.2A priority patent/EP3480215B8/en
Priority to BR112016002752-3A priority patent/BR112016002752B1/pt
Priority to RU2016106339A priority patent/RU2656161C1/ru
Priority to US15/037,104 priority patent/US10087260B2/en
Priority to CA2919359A priority patent/CA2919359C/en
Priority to JP2016537121A priority patent/JP6326137B2/ja
Priority to CN201811053363.6A priority patent/CN110240655B/zh
Application filed by 烟台荣昌生物工程有限公司 filed Critical 烟台荣昌生物工程有限公司
Priority to AU2014352475A priority patent/AU2014352475B2/en
Priority to DK14864053.5T priority patent/DK3072907T3/en
Priority to EP14864053.5A priority patent/EP3072907B1/en
Priority to PL18204950T priority patent/PL3480215T3/pl
Priority to CN201480006648.8A priority patent/CN105008398B/zh
Priority to KR1020187012035A priority patent/KR101936697B1/ko
Priority to KR1020187036881A priority patent/KR101993136B1/ko
Publication of WO2015074528A1 publication Critical patent/WO2015074528A1/zh
Priority to CY20211100704T priority patent/CY1124651T1/el

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2896Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against molecules with a "CD"-designation, not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/6805Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a vinca alkaloid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • A61K47/6855Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell the tumour determinant being from breast cancer cell
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the present invention relates to novel HER2 antibodies or functional fragments thereof comprising engineered heavy and light chains.
  • the invention also relates to the improved conjugate of a HER2 antibody and a small molecule drug.
  • the invention also relates to the use of the antibody and the conjugate in the manufacture of a medicament for the treatment of a tumor.
  • ErbB2 also known as HER2/neu
  • HER2/neu is the second member of the EGFR family, and ErbB2 exerts a biological role by forming a heterodimer with the other three members of the EGFR family. No ligands that bind directly to ErbB2 have been found.
  • the neu gene encoding ErbB2 was first isolated from rat neuroblastoma.
  • the homologous gene of the neu gene in human somatic cells, called HER2 is located on the long arm of chromosome 17 (17q21.1), and the encoded product ErbB2 is composed of 1255 amino acids with a molecular weight of about 185kDa, of which 720-987 belong to the tyrosine. Aminokinase kinase active domain.
  • ErbB2 can also reduce the expression of cyclin D and c-myc, thereby reducing the expression of cyclin-dependent kinase (cdk) inhibitor p27kipl, cdk2 The activity is inhibited and leads to cell proliferation [1].
  • HER2 was found to be expressed and overexpressed in various tumors.
  • the positive expression, overexpression and HER2 overexpression of HER2 in several tumors have been reported as follows: ovarian cancer 45 %, 21%, 23,316 [2]; breast cancer 58%, 38%, 223,112 people [3]. Therefore, there is a clinical need for effective drugs targeting HER2 to treat malignant tumors.
  • HER2-targeted monoclonal antibodies have trastuzumab and pertuzumab.
  • Trastuzumab is a humanized monoclonal antibody directed against HER2 developed by Genentech, USA.
  • Trastuzumab not only has a high affinity for the HER2 receptor, but also solves the problem of the immunogenicity of murine antibodies applied to humans.
  • Clinical trial results show that the effective rate of Trastuzumab alone is 11.6% to 16%, and the effective rate of combination with chemical drugs can reach 50%. Patients with advanced recurrent breast cancer have longer survival and lower mortality than chemotherapy alone.
  • pertuzumab Another antibody against HER2 is pertuzumab [4], which was also developed by Genentech, USA, and pertuzumab binds to the extracellular domain II region of the HER2 receptor. Inhibition of the formation of dimers, thereby inhibiting receptor-mediated signal transduction pathways.
  • Trastuzumab (Herceptin) binds to the extracellular IV region of the HER-2 receptor. The US FDA approved pertuzumab on June 8, 2012 for the treatment of HER2-positive patients with advanced metastatic breast cancer (see CN101023100B).
  • Monoclonal antibody therapy has attracted more and more attention due to its high target specificity and low side effects, but its efficacy is limited when used alone.
  • anti-tumor monoclonal antibodies are most successful against lymphocyte tumors, such as non-Hawking's (NHL), chronic lymphocyte tumors.
  • NHL non-Hawking's
  • Rituxan's Phase II clinical study of NHL showed only 6% complete response. There is also a 15% response to metastatic breast cancer. Therefore, most monoclonal antibodies are used in combination with chemotherapy.
  • Rituxan can be used in combination with standard chemotherapy to treat chronic lymphocytic tumors with an efficiency that can be increased to 90%.
  • the main route to improve the efficacy of monoclonal antibodies is antibody drug conjugates.
  • Antibody drug conjugates belong to a new class of anti-cancer biological missile drugs, which are composed of three parts: antibodies, cytotoxics and linkers connecting the two.
  • Chemical Coupling After coupling a monoclonal antibody to a cytotoxin, the antibody drug conjugate utilizes the targeting of the monoclonal antibody to specifically recognize the receptor on the surface of the cancer cell, bind to the receptor, and then enter the cell interior.
  • the use of intracellular proteases to release cellular toxicants prevents cancer cells from multiplying and killing cancer cells.
  • the antibody drug coupling technology integrates small molecule drugs with biological proteins, which combines the advantages of both, greatly enhances the efficacy, reduces toxic side effects, and becomes a new generation of therapeutic products.
  • the first clinically successful example of targeting antibody drug conjugates is Gemtuzumab ozogamicin (Wyeth trade name Mylotarg).
  • Mylotarg is the first monoclonal antibody to be marketed. This medicine is made up of anti-CD33 antibody, DNA degradation drug Calicheamicin is composed of the chemical connector AcBut.
  • Mylotarg is a humanized anti-CD33 IgG4 coupled with the anticancer drug Calicheamicin for the treatment of acute myeloid leukemia [5].
  • Mylotarg is the first generation of monoclonal antibody-conjugated drug. There are three fatal defects in the technology. First, the linker used to connect the poison is very unstable. The half-life is only 2 days. The poison is seriously shed and the clinical side shows high toxicity.
  • the antibody is coupled to the linker by the amino group of lysine, and there are dozens of lysines on the surface of an antibody.
  • the coupling sites are random, partially affecting the efficacy, and more importantly, the coupling technology at the time. Immature, only 50% of the antibodies are conjugated, and the clinical efficacy is not ideal.
  • the antibody used is IgG4, lacking antibody-mediated cytotoxicity (ADCC) and complement-mediated cytotoxicity ( CDC). Therefore, after 10 years of listing, Mylotarg was withdrawn due to its toxic side effects and limited efficacy.
  • a second clinically successful example of targeting antibody drug conjugates is a new drug for the treatment of Hodgkin's lymphoma.
  • the drug was only used in Phase II clinical trials, the US FDA approved its listing in 2011 due to its particularly good efficacy.
  • the drug is a novel targeted antibody-drug conjugate (ADC) developed by Seattle Genetics to target two types of lymphoma patients expressing CD30 antigen.
  • ADC antibody-drug conjugate
  • berentuximab is composed of an anti-CD30 monoclonal antibody, a microtubule inhibitor (MMAE) and a dipeptide chemical linker.
  • MMAE microtubule inhibitor
  • the antibody drug conjugate has the characteristics of low side effects and high efficiency of inhibiting lymphoma.
  • T-DM1 for anti-malignant breast cancer developed by Genentech Inc.
  • the monoclonal antibody to the antibody drug conjugate is HER2 (ErbB2) on the surface of breast cancer cells, and the conjugated cytotoxicity is microtubule inhibitor DM1.
  • HER2 ErbB2
  • the phase III clinical trial results show that the results are better than chemotherapy and have fewer side effects.
  • Breast cancer patients have previously been treated with Herceptin and taxane chemotherapy, but their condition is still progressing, but they are treated with antibody drug conjugates. Patients can significantly prolong the survival of patients with HER2-positive breast cancer without deterioration of the disease [9].
  • the US Food and Drug Administration (FDA) approved the drug on February 22, 2013 for the treatment of patients with HER2-positive advanced metastatic breast cancer (see CN100482281C).
  • Herceptin is a breakthrough in the history of treatment of HER2 overexpressing breast cancer that has tried multiple anticancer therapies, about 85% of subjects have no or only weak response to Herceptin therapy [11]. Studies have shown that HER2 is expressed and overexpressed in a variety of tumors, so there is a clinical need to develop HER2-targeted anticancer drugs for HER2 overexpressing tumors that have no or only weak response to Herceptin therapy. Or other patients with disease related to HER2 expression (not just breast cancer).
  • the present invention provides a solution to meet this need.
  • the invention provides an antibody or functional fragment thereof that is capable of specifically binding to HER2.
  • the antibody comprises a heavy chain and a light chain, wherein
  • the heavy chain comprises three CDR regions, wherein the amino acid sequence of at least one of the CDR regions has an amino acid sequence as set forth in SEQ ID NO: 1, 2 or 3 or has at least 80% (preferably 85%) , 90%, 95%, 98%, or 99%) sequences of sequence identity;
  • the light chain comprises three CDR regions, wherein the amino acid sequence of at least one of the CDR regions has or has at least 80% (preferably 85%) of the amino acid sequence set forth in SEQ ID NO: 4, 5 or , 90%, 95%, 98%, or 99%) sequences of sequence identity.
  • the antibody comprises a heavy chain and a light chain, wherein
  • the heavy chain comprises at least three CDR regions having amino acid sequences as set forth in SEQ ID NOs: 1, 2 and 3, respectively;
  • the light chain comprises at least three CDR regions having the amino acid sequences set forth in SEQ ID NOs: 4, 5 and 6, respectively.
  • the invention is provided on August 22, 2013 Deposit No. 8102 (CGMCC No. 8102) was deposited at the General Microbiology Center of the China Microbial Culture Collection Management Committee (Microbiology Institute, Chinese Academy of Sciences, No. 3, Beichen West Road, Chaoyang District, Beijing, China, 100101) Secreted antibodies or antibodies derived from them (translated to the deposit of the Budapest Treaty on October 29, 2013).
  • the present invention provides deposits with the China Center for Type Culture Collections on November 06, 2013 under the accession number C2013170 (CCTCC C2013170) (Wuhan University, Wuchang, Wuhan, Hubei province, China, 430072) An antibody secreted by Chinese hamster ovary cells (CHO cells) or an antibody derived therefrom.
  • CTCC C2013170 Chinese hamster ovary cells
  • the invention provides an isolated polynucleotide encoding an antibody of the invention.
  • the invention provides a combination of isolated polynucleotides comprising a polynucleotide encoding a light chain of an antibody of the invention or a functional fragment thereof, and a heavy chain encoding an antibody of the invention or a functional fragment thereof Polynucleotide.
  • the invention provides an expression vector comprising a polynucleotide according to the invention or a combination of polynucleotides according to the invention, said polynucleotide and a polypeptide which allows it to be encoded in a host cell or cell-free
  • the regulatory sequences expressed in the expression system are operably linked.
  • the invention provides a conjugate comprising an antibody of the invention or a functional fragment thereof, conjugated to one or more therapeutic agents, preferably the therapeutic agent is a cytotoxic drug (eg An antimetabolite, an antitumor antibiotic, an alkaloid), an immunopotentiator or a radioisotope, more preferably the therapeutic agent is selected from maytansinoids (eg, Ansamitocin or Mertansine) )), dolastatin and derivatives thereof, most preferably the therapeutic agent is selected from the group consisting of MMAE (Monomethyl auristatin E, monomethyl auristatin peptide E) and MMAF (Monomethyl auristatin F, monomethyl ear) Inhibin peptide F).
  • the therapeutic agent can also be selected from those listed in Table 1 below.
  • MMAE Monomethyl auristatin E Microtubule monomeric protein polymerization inhibitor
  • MMAE derivative Monomethyl auristatin F Microtubule monomeric protein polymerization inhibitor
  • MMAF derivative DM1 Mertansine derivative M4 Microtubule-depolymerizing [15] DM4 Mertansine derivative M4 Microtubule depolymerization [15] Duocarmycine Duocarmycine DNA conjugate [13] Calicheamicin Calicheamicin DNA minor groove conjugate [13] PBDA Pyrrolobenzodiazepines DNA conjugate [13] Doxorubicin Doxorubicin Topoisomerase inhibitors [13] Vinca Alkaloids Vinca Alkaloids [13] Metrotrexate Metrotrexate [13] Vinblastine Vinblastine Microtubule depolymerization [13] Daunorubicin Daunorubicin [13]
  • the therapeutic agent is coupled to the antibody or a functional fragment thereof via a linker.
  • the linkers used in the present invention can be attached to the antibody by any means known in the art, preferably via a thiol group and/or an amino group.
  • the antibodies of the invention are linked to the linker via a thiol group.
  • the joint used in the present invention may be a cleavable joint (i.e., a joint that can be broken in an in vivo environment) or a non-cleavable joint.
  • the linker of the invention is selected from a cleavable linker, preferably selected from the group consisting of a peptide, a guanidine, and a disulfide linker, such as a maleimidocaproyl-valine-citrulline-p-amino group.
  • a cleavable linker preferably selected from the group consisting of a peptide, a guanidine, and a disulfide linker, such as a maleimidocaproyl-valine-citrulline-p-amino group.
  • benzyloxycarbonyl hereinafter abbreviated as mc-vc-pAB or vc, ie, maleimidocaproyl-valine-citrulline-p-aminobenzyloxycarbonyl.
  • the linkers of the invention are selected from non-cleavable linkers, such as maleimidocaproyl (hereinafter abbreviated as mc, ie, maleimidocaproyl).
  • the linker can also be selected from those listed in Table 2 below.
  • the invention provides a conjugate having the general formula Ab-(LU) n , wherein Ab represents an antibody or functional fragment thereof according to the invention, and L represents a linker (eg mc-vc-pAB or mc)
  • U represents a therapeutic agent (preferably the therapeutic agent is selected from the group consisting of a cytotoxic drug, an immunopotentiator, and a radioisotope, and more preferably the therapeutic agent is selected from the group consisting of maytansinoids, dolastatin peptides and derivatives thereof Most preferably the therapeutic agent is selected from the group consisting of MMAE and MMAF), and n is an integer from 1 to 8 (eg 1, 2, 3, 4, 5, 6, 7, or 8).
  • the joint used in the present invention may be a cleavable joint (i.e., a joint that can be broken in an in vivo environment) or a non-cleavable joint.
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising an antibody or functional fragment thereof according to the invention and/or a conjugate according to the invention, and a pharmaceutically acceptable carrier.
  • the invention provides a method of treating or preventing cancer, in particular a HER2-positive cancer, comprising administering a therapeutically effective amount to a subject in need thereof
  • An antibody, polynucleotide, polynucleotide combination, expression vector, conjugate and/or pharmaceutical composition according to the invention comprising administering a therapeutically effective amount to a subject in need thereof
  • the invention provides the use of an antibody, polynucleotide, polynucleotide combination, expression vector, conjugate, and/or pharmaceutical composition according to the invention in the manufacture of a medicament for the treatment or prevention of cancer.
  • the invention provides an antibody, polynucleotide, polynucleotide combination, expression vector, conjugate, and/or pharmaceutical composition according to the invention for use in the treatment or prevention of cancer.
  • the cancer is a HER2-positive cancer, more preferably selected from breast cancer, ovarian cancer or gastric cancer. More preferably, the cancer is lapatinib and/or Herceptin resistant cancer, such as lapatinib and/or Herceptin resistant breast, ovarian or gastric cancer.
  • the present invention provides a hybridoma cell deposited with the General Microbiology Center of the Chinese Collection of Microorganisms and Cultures on August 22, 2013 under the accession number No. 8102 (domestic deposit was transferred to the Budapest Treaty) The date is October 29, 2013).
  • the present invention provides a CHO cell deposited with the China Center for Type Culture Collection on November 06, 2013 under the accession number C2013170.
  • the invention relates to antibody-antibody-drug conjugates that are capable of treating cancer.
  • the conjugate comprises a monoclonal antibody capable of specifically binding to a cancer cell surface receptor, a small molecule drug having a cytotoxic effect, and a linker capable of linking the above two parts together by a covalent bond.
  • the invention also relates to the use of these conjugates in the manufacture of a medicament for the treatment of breast cancer and/or ovarian cancer and/or gastric cancer.
  • the present invention relates to antibody-small molecule drug conjugates having the general formula Ab-(LU) n , Ab representing a monoclonal antibody that targets HER2, and L being selected from mc-vc-pAB Or mc, U is selected from MMAE or MMAF, and n is an integer from 1 to 8.
  • the humanized antibody targeting HER2 disclosed herein is RC48, and the amino acid sequences of the heavy chain CDRs region thereof are represented by SEQ ID NO. 1, SEQ ID NO. 2 and SEQ ID NO.
  • the humanized antibody targeting HER2 disclosed in the present invention is RC48,
  • the amino acid sequences of the light chain CDRs region are represented by SEQ ID NO. 4, SEQ ID NO. 5 and SEQ ID NO. 6, respectively.
  • the above-described HER2-targeted humanized antibody disclosed in the present invention is RC48, which is secreted by a deposited cell deposited with the China Center for Type Culture Collection under the accession number C2013170 on November 6, 2013.
  • Trastuzumab It is a recombinant humanized monoclonal antibody that selectively acts on the extracellular domain of human epidermal growth factor receptor-2 (HER2) and is mainly used to treat HER2-positive cancers.
  • the humanized antibody RC48 of the present invention is a recombinant human HER2 antibody capable of binding to the extracellular domain of HER2 with high affinity. In vitro and in vivo, RC48 monoclonal antibody shows a person capable of inhibiting overexpression of HER2. Tumor cell proliferation.
  • the two small molecule cytotoxic agents involved in the present invention are MMAE (Monomethyl auristatin E, monomethyl auristatin peptide E) or MMAF (Monomethyl auristatin F, monomethyl auristatin peptide F) (see Figure 6). It is the inhibition of small molecules by two kinds of cell tubulin.
  • the present invention also relates to two linker maleimidocaproyl (hereinafter abbreviated as mc) and maleimido-hexanoyl-valine-citrulline-p-aminobenzyloxy ( Maleimido-Caproyl-Valine-Citrulline-p-AminoBenzyloxy) (hereinafter abbreviated as mc-vc-pAB) (see Figure 7), which can also be simply represented by vc in the name of the conjugate, the former being non-cleavable, The latter is cleavable and the corresponding conjugate exhibits different stability and half-life in vivo.
  • mc-vc-pAB linker maleimidocaproyl
  • mc-vc-pAB Maleimido-hexanoyl-valine-citrulline-p-aminobenzyloxy
  • mc-vc-pAB Maleimido-Caproyl-Valine-Citrulline
  • RC48 mAb is linked to the linker by cysteine to form the following three antibody drug conjugates RC48-vc-MMAE (see Figure 8), RC48-vc-MMAF (see Figure 9) and RC48-mc-MMAF (see Figure 9). See Figure 10).
  • the in vitro antigen-antibody binding ability of the conjugates involved in the present invention is comparable to that of the RC48 naked antibody and T-DM1; in the cell activity assay, the cytotoxic activity is significantly higher than that of the RC48 naked antibody, Herceptin, T-DM1, and the experimental cells include HER2.
  • the conjugate of the present invention has a significant antitumor effect on BT474 human breast cancer-bearing nude mice (Fig.
  • the rapamycin-resistant tumor-bearing nude mice also showed significant anti-tumor activity, and the effect was significantly better than that of the positive control drug (Fig. 15). Meanwhile, the antibody conjugate of the present invention inhibited tumor growth of ovarian cancer and gastric cancer. Nude mice also showed unexpected anti-tumor effects ( Figures 16 and 17).
  • the maximum tolerated dose of the conjugate of the present invention was determined by in vivo experiments in mice to be RC48-mc-MMAF: >150 mg/kg, RC48-vc-MMAF: 60 mg/kg, RC48-vc-MMAE: 100 mg/kg further.
  • the human ovarian cancer xenograft model was used to test the effect of the tumor.
  • the volume of the mouse tumor model and the body weight of the mouse were observed.
  • the conjugate effect was significantly higher than that of the naked antibody and T-DM1.
  • the weight gain of the mice (Fig. 18) showed good efficacy with low toxicity and high efficiency.
  • the conjugate provides a new drug candidate for the treatment of HER2-positive cancer, HER2 antibody drug-resistant cancer, tyrosine kinase inhibitor-resistant cancer and other related diseases.
  • an antibody or functional fragment thereof according to the invention is isolated.
  • an antibody or functional fragment thereof according to the invention is a monoclonal antibody.
  • an antibody or functional fragment thereof according to the invention is a humanized antibody.
  • an antibody or functional fragment thereof according to the invention has ADCC activity.
  • an antibody or functional fragment thereof according to the invention has CDC activity.
  • an antibody or functional fragment thereof according to the invention specifically binds to HER2 without substantially binding to EGFR, HER3 and HER4.
  • the antibody or functional fragment thereof according to the invention is an IgG1 kappa antibody.
  • an antibody or functional fragment thereof according to the invention can be used to treat or prevent cancer, wherein the cancer overexpresses HER2.
  • Figure 1 is a SDS-PAGE image of purified human recombinant protein HER2-ECD stained with Coomassie brilliant blue. The loading amount per well was 10 ⁇ g.
  • Figure 2 shows the SDS-PAGE analysis of cRC48, RC48, with an antibody loading of 2 ⁇ g per well.
  • Figure 3 shows the binding affinity of HER2-ECD of the humanized antibody RC48 as determined by ELISA assay, and the binding affinity constant Kd was calculated. Herceptin and cRC48 were used as controls in this experiment.
  • Figure 4A shows the ability of anti-HER2 humanized antibody RC48 to bind to HER2 + cells SK-BR3, BT474, HER2 - cell MDA-MB468 by flow cytometry.
  • Figure 4B shows the ability of flow cytometry to analyze the binding of anti-HER2 antibodies to BT474 cell surface antigens at various antibody concentrations.
  • Anti-HER2 antibodies include Herceptin, cRC48, RC48. A total of 5 ⁇ 10 4 cells were analyzed.
  • Figure 5 shows that RC48 only shows specific binding affinity for HER2, but no binding to EGFR, HER3, HER4.
  • Figure 6 shows the molecular structure of tubulin conjugates MMAE and MMAF.
  • Figure 7 shows the molecular structure of the chemical linkers mc-vc-pAB and mc.
  • Figure 8 shows the molecular structure of the RC48 antibody drug conjugate (RC48-vc-MMAE).
  • Figure 9 shows the molecular structure of the RC48 antibody drug conjugate (RC48-vc-MMAF).
  • Figure 10 shows the molecular structure of the RC48 antibody drug conjugate (RC48-mc-MMAF).
  • Figure 11 shows the anti-tumor effect of RC48 on the BT474 human breast cancer suppressor model.
  • Figure 12 shows the growth inhibitory effect of the RC48 conjugate on HER2-positive cells SK-BR-3.
  • Figure 13 shows the growth inhibitory effect of the RC48 conjugate on HER2-positive cells SK-OV-3.
  • Figure 14 shows the anti-tumor effect of RC48 conjugate on the BT474 human breast cancer-bearing nude mouse model.
  • Figure 15 shows the efficacy of RC48-vc-MMAE, T-DM1 on Herceptin and Lapatinib-resistant human breast cancer BT-474/L1.9 nude mice xenografts.
  • Figure 16 shows the RC48 conjugate against the SK-OV-3 human ovarian cancer suppressor model. Antitumor effect.
  • Figure 17 shows the efficacy of RC48-vc-MMAE, Herceptin, and lapatinib on human gastric cancer NCI-N87 nude mice xenografts.
  • Figure 18 shows the effect of different antibody drug conjugates on body weight of mice.
  • compositions are used interchangeably and mean at least one drug, and optionally a pharmaceutically acceptable carrier, that are combined together to achieve a particular purpose or A combination of excipients.
  • the pharmaceutical compositions include combinations that are separated in time and/or space, as long as they are capable of acting together to achieve the objectives of the present invention.
  • the components contained in the pharmaceutical composition eg, antibodies, nucleic acid molecules, nucleic acid molecule combinations, and/or conjugates according to the invention
  • the components contained in the pharmaceutical composition can be administered to the subject as a whole or separately to the subject.
  • the components contained in the pharmaceutical composition When the components contained in the pharmaceutical composition are When applied to a subject, the components can be administered to the subject simultaneously or sequentially.
  • the pharmaceutically acceptable carrier is water, a buffered aqueous solution, an isotonic saline solution such as PBS (phosphate buffer), dextrose, mannitol, dextrose, lactose, starch, magnesium stearate, cellulose, carbonic acid.
  • PBS phosphate buffer
  • the type of pharmaceutically acceptable carrier employed depends inter alia on whether the composition according to the invention is formulated for oral, nasal, intradermal, subcutaneous, intramuscular or intravenous administration.
  • the composition according to the invention may comprise a wetting agent, an emulsifier or a buffer substance as an additive.
  • compositions, vaccine or pharmaceutical preparation according to the invention may be administered by any suitable route, for example, orally, nasally, intradermally, subcutaneously, intramuscularly or intravenously.
  • therapeutic agent denotes any substance or entity capable of exerting a therapeutic effect (eg, treating, preventing, ameliorating or inhibiting any disease and/or condition), including but not limited to: chemotherapeutic agents, radiation A therapeutic agent, an immunotherapeutic agent, a thermally therapeutic agent, and the like.
  • CDR region refers to the hypervariable region of the heavy and light chains of an immunoglobulin, as defined by Kabat et al. (Kabat et al., Sequences of proteins of immunological interest, 5th Ed. , USDepartment of Health and Human Services, NIH, 1991, and later). There are three heavy chain CDRs and three light chain CDRs. The term CDR or CDRs as used herein is used to indicate one of these regions, or a few or even all of these regions, which contain a majority of the amino acid residues responsible for binding by the affinity of the antibody for the antigen or its recognition epitope. base.
  • identity refers to the two to be compared after the optimal alignment (optimal alignment).
  • the percentage of identical nucleotides or identical amino acid residues between the sequences which are purely statistical and the differences between the two sequences are randomly distributed and cover the full length thereof.
  • Sequence comparisons between two nucleic acid or amino acid sequences are typically performed by comparing the sequences after they have been optimally matched, which can be performed by segments or by a "comparison window".
  • the optimal alignment for comparing sequences can also be achieved by the local homology algorithm of Smith and Waterman (1981) [Ad. App. Math.
  • terapéuticaally effective amount refers to a dose sufficient to demonstrate its benefit to the subject to which it is administered.
  • the actual amount administered, as well as the rate and time course of administration, will depend on the condition and severity of the subject being treated.
  • the prescription for treatment eg, the determination of the dose, etc.
  • the prescription for treatment is ultimately the responsibility of the GP and other physicians and depends on their decision, usually considering the disease being treated, the condition of the individual patient, the site of delivery, the method of administration, and the Other factors known.
  • subject refers to a mammal, such as a human, but may also be other animals, such as wild animals (such as herons, donkeys, cranes, etc.), livestock (such as ducks, geese, etc.) or experimental animals (such as Orangutans, monkeys, rats, mice, rabbits, guinea pigs, woodchucks, ground squirrels, etc.).
  • wild animals such as herons, donkeys, cranes, etc.
  • livestock such as ducks, geese, etc.
  • experimental animals such as Orangutans, monkeys, rats, mice, rabbits, guinea pigs, woodchucks, ground squirrels, etc.
  • antibody refers to an intact antibody and any antigen-binding fragment thereof ("antigen-binding portion") or single strand thereof.
  • Fral length antibody refers to a protein comprising at least two heavy (H) chains and two light (L) chains interconnected by a disulfide bond.
  • Each heavy chain comprises a heavy chain variable region (abbreviated as VH) and a heavy chain constant region.
  • the heavy chain constant region comprises three domains, CH1, CH2 and CH3.
  • Each light chain comprises a light chain variable region (abbreviated as VL) and a light chain constant region.
  • the light chain constant region contains a domain, CL.
  • VH and VL regions can also be subdivided into multiple regions of high variability, referred to as complementarity determining regions (CDRs), interspersed with more conserved regions called framework regions (FR).
  • CDRs complementarity determining regions
  • FR framework regions
  • Each VH and VL consists of three CDRs and four FRs, arranged from amino terminus to carboxy terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • These variable regions of the heavy and light chains comprise a binding domain that interacts with the antigen.
  • the constant region of the antibody mediates binding of the immunoglobulin to the host's tissues or factors, including various cells of the immune system (such as effector cells) and the first component (Clq) of the classical complement system.
  • Chimeric or humanized antibodies are also encompassed in the antibodies according to the invention.
  • humanized antibody refers to an antibody comprising a CDR region derived from a non-human antibody, and the other portion of the antibody molecule is derived from one (or several) human antibodies. Moreover, in order to retain binding affinity, some residues of the backbone (referred to as FR) segment can be modified (Jones et al., Nature, 321:522-525, 1986; Verhoeyen et al., Science, 239: 1534-1536, 1988; Riechmann et al., Nature, 332: 323-327, 1988). Humanized antibodies or fragments thereof according to the invention can be prepared by techniques known to those skilled in the art (for example, as described in the document Singer et al., J. Immun. 150: 2844-2857, 1992; Mountain et al., Biotechnol. Genet. Eng. Rev., 10: 1-142, 1992; or Bebbington et al., Bio/Technology, 10: 169-175, 1992).
  • chimeric antibody refers to an antibody wherein the variable region sequence is from one species and the constant region sequence is from another species, eg, the variable region sequence is derived from a mouse antibody and the constant region sequence is derived from a human antibody.
  • a chimeric antibody or fragment thereof according to the invention can be prepared by using genetic recombination techniques.
  • the chimeric antibody can be produced by cloning recombinant DNA comprising a promoter and a sequence encoding a variable region of a non-human, in particular murine, monoclonal antibody according to the invention, and a sequence encoding a constant region of a human antibody .
  • the chimeric antibody of the present invention encoded by such a recombinant gene will be, for example, a murine-human chimera whose specificity is determined by a variable region derived from murine DNA, and whose isotype is derived from human DNA. Constant zone to determine.
  • a murine-human chimera whose specificity is determined by a variable region derived from murine DNA, and whose isotype is derived from human DNA. Constant zone to determine.
  • Verhoeyn et al. BioEssays, 8: 74, 1988.
  • monoclonal antibody refers to a preparation of an antibody molecule having a single molecular composition. Monoclonal antibody compositions display a single binding specificity and affinity for a particular epitope.
  • mRC48 antibody refers to the anti-HER2 murine monoclonal antibody mRC48 obtained by the present inventors.
  • RC48 antibody refers to a humanized anti-HER2 antibody RC48 which is derived from the mRC48 antibody by humanization.
  • the cRC48 antibody referred to herein refers to a chimeric RC48 antibody, ie, a human-mouse chimeric antibody, comprising a murine variable region and a human constant region.
  • the cRC48 antibody differs from the RC48 antibody only in the difference in the framework regions in the variable region, the framework region of cRC48 is murine, and the framework region of RC48 is human.
  • the term "functional fragment” as used herein especially refers to antibody fragments such as Fv, scFv (sc refers to single strand), Fab, F(ab')2, Fab', scFv-Fc fragment or double A diabody, or any fragment that is capable of increasing half-life by chemical modification or by incorporation into a liposome, such as the addition of a poly(alkylene) glycol such as polyethylene glycol (“poly” Glycolation, PEGylation”) (called PEGylated fragment called Fv-PEG, scFv-PEG, Fab-PEG, F(ab')2-PEG or Fab'-PEG) ("PEG” is poly(ethylene) Glycol), the fragment has EGFR binding activity.
  • a poly(alkylene) glycol such as polyethylene glycol (“poly” Glycolation, PEGylation")
  • PEGylated fragment called Fv-PEG, scFv-PEG, Fab
  • the functional fragment will consist of or comprise a partial sequence of a heavy or light variable chain from which the antibody is derived, the partial sequence being sufficient to retain the same binding specificity and sufficient affinity as the antibody from which it is derived, preferably at least for EGFR It is equal to 1/100 of the affinity of the antibody from which it is derived, and in a more preferred manner is at least equal to 1/10.
  • a functional fragment will comprise a minimum of 5 amino acids, preferably 10, 15, 25, 50 and 100 contiguous amino acids of the antibody sequence from which it is derived.
  • the monoclonal antibody according to the invention may, for example, be purified on an affinity column, to which a HER2 antigen (e.g., HER2-ECD) has been immobilized on the affinity column or which comprises a monoclonal antibody specific according to the present invention.
  • a HER2 antigen e.g., HER2-ECD
  • the monoclonal antibody can be purified by protein A and/or G chromatography, with or without the purpose of eliminating residual protein contaminants and ion exchange chromatography of DNA and LPS, either alone or not. Exclusion chromatography on Sepharose gels to eliminate potential aggregates due to the presence of dimers or other multimers. In a more preferred manner, all of these techniques can be used simultaneously or continuously.
  • dolastatin refers to a polypeptide isolated from a marine truncated sea rabbit (Dollabella auricularia) including, but not limited to, dolastatin 10 and dolastatin. Peptide 15 (dolastatin 15).
  • the dolastatin peptide is a mitotic inhibitor which exhibits strong anticancer activity and is therefore candidates for anticancer drugs.
  • researchers have further discovered and synthesized a number of derivatives of the dolastatin peptide, such as MMAE and MMAF.
  • linker refers to the portion of an antibody drug conjugate (ie, ADC) that attaches an antibody to a drug, which may be cleavable or non-cleavable.
  • a cleavable linker ie, a cleavable linker or a biodegradable linker
  • the linkers of the present invention have very good stability, greatly reducing the release of the drug during delivery to the target (eg, in the blood), thereby reducing side effects and toxicity.
  • the linker of the invention is selected from a cleavable linker, such as a disulfide-based linker (which selectively cleaves in tumor cells having a higher concentration of sulfhydryl groups), a peptide linker (which is in a tumor cell) Enzyme cut), ⁇ joint.
  • the linkers of the invention are selected from non-cleavable linkers, such as thioether linkers.
  • the linker of the invention is selected from the group consisting of a cleavable mc-vc-pAB linker and a non-cleavable mc linker.
  • HER2-ECD composed of the extracellular region of HER2 (ECD) was prepared, which was used as an antigen for subsequent immunoreaction, production of monoclonal antibodies, and various biological assays.
  • a cDNA fragment encoding HER2-ECD (amino acid Thr23 to Thr652, GenBank Accession No. M11730) was cloned into a pcDNA3 (Invitrogen) expression vector by PCR.
  • the cDNA of the HER2-ECD coding region was obtained from the HER2 + SKBR3 cell line (ATCC No.: HTB-30) by RT-PCR (the kit was subjected to Promega's ImProm-IITM Reverse Transcription System reverse transcription system).
  • the cDNA of HER2-ECD was used as a template for PCR amplification using the above primers.
  • the amplification conditions were: denaturation at 94 ° C for 30 s, annealing at 60 ° C for 30 s, extension at 72 ° C for 1 minute, 30 cycles, and finally extension at 72 ° C for 10 minutes.
  • the PCR fragment was then recovered, digested with BamHI and XbaI enzyme (NEB), and ligated with the pcDNA3 vector.
  • a poly-histidine tag was added to the C-terminus of HER2-ECD to facilitate purification.
  • HEK293 cells ATCC, USA
  • Monoclonal antibodies were prepared by immunizing mice with the HER2-ECD prepared above as an antigen.
  • the immune response, hybridoma cell fusion, and primary screening were performed according to standard procedures (Reference: WHO Technical Report Series, No. 822, 1992 Annex 3).
  • 0.25ml of HER2-ECD protein (50-100 ⁇ g) and 0.25ml of Freund's complete adjuvant (Difco Lab) were mixed in equal volume to immunize 4 Balb/c mice (purchased from Shanghai Slack Laboratory Animals Co., Ltd.), at intervals After 2 weeks, the second injection was performed, using Freund's incomplete adjuvant (Difco Lab), the antigen amount was 25-50 ⁇ g/0.5 ml/mouse, and the third injection was performed after 3 weeks.
  • the injection dose was the same as the second.
  • the blood was taken 10 days after the third injection.
  • the serum of the mice was detected by an enzyme-linked immunosorbent assay (ELISA), and the spleens of the two mice with the highest anti-HER2 antibody titer in the serum were taken out, and then fused with myeloma cells P3X63Ag8 (ATCC CRL-1580).
  • the fused cells were diluted into 10 96-well plates and screened by ELISA according to the binding ability to HER2-ECD.
  • HER2-ECD HER2-ECD (0.2-1 ⁇ g/ml) and then incubated with gradient-diluted mouse serum or hybridoma supernatant (100 ⁇ L).
  • the murine anti-HER2 antibody was detected with a horseradish peroxidase-conjugated goat F(ab') 2 anti-mouse IgG Fc specific secondary antibody (Invitrogen).
  • the supernatants of 400 hybridoma cell lines were screened by ELISA, and 36 of them showed strong HER2-ECD binding.
  • Ten hybridoma cells with the strongest HER2 binding ability were selected, and the subcloned hybridoma cell lines were screened by limiting dilution method.
  • Hybrid subcloning of hybridoma cell lines, protein purification, determination of HER2 binding by ELISA Affinity, flow cytometry (BD FACS Calibur) was used to further test their ability to bind to HER2 naturally expressed on the surface of human breast cancer cell lines (see Example 4 for a more detailed description).
  • hybridoma cell line mRC48 (murine-derived IgG1k) was identified by sequence analysis, which has strong HER2 binding ability, and was further analyzed by ELISA and cell assay.
  • the hybridoma cell mRC48 was deposited with the General Microbiology Center of the China Microbial Culture Collection Management Committee on August 22, 2013 under the accession number No. 8102 (the date of transfer to the Budapest Treaty was October 29, 2013).
  • variable regions of the above hybridoma cell clone mRC48 heavy and light chains were sequenced by rapid amplification of the 5' end using a commercial kit SMARTTM RACE cDNA Amplification Kit (Clontech) according to the instructions.
  • RNApure Tissue Kit (Conway Beijing Century Biotech Co., Ltd.) Total RNA was extracted from hybridoma cells using SMART TM RACE cDNA Amplification Kit Total RNA reverse transcription, total RNA as a template, primers kit Add the reverse transcriptase SMARTScribeTM Reverse Transcriptase, perform reverse transcription according to the procedure provided in the kit to obtain RACE-Ready first strand cDNA, and then perform two rounds of PCR. The first round of PCR can be used to obtain the cDNA as a template, which is provided in the kit.
  • the UPM is the 5' primer and the 3' primer is mRC48-VL-1/mRC48-VH-1.
  • the PCR reaction conditions were: pre-denaturation at 94 ° C for 5 min; 25 amplification cycles (denaturation at 94 ° C for 30 s, annealing at 68 ° C for 30 s, extension at 72 ° C for 2 min); and finally extension at 72 ° C for 10 min.
  • the product of the first round of PCR was used as a template, and the NUP provided in the kit was a 5' primer and the 3' primer was mRC48-VL-2/mRC48-VH-2.
  • the PCR reaction conditions were: Pre-denaturation at 94 ° C for 5 min; 25 cycles of amplification (denaturation at 94 ° C for 30 s, annealing at 68 ° C for 30 s, extension at 72 ° C for 2 min); extension at 72 ° C for 10 min.
  • both the variable regions of the heavy and light chains of the hybridoma cell clone mRC48 were obtained.
  • the PCR product was purified by agarose gel electrophoresis and subcloned into the pCR2.1 TOPO cloning vector (Invitrogen). Plasmid DNA of 10 independent clones was obtained by PCR and sequenced with M13 forward and reverse primers. DNA sequence analysis indicated that all 10 clones had cDNA encoding the same VH or VL polypeptide.
  • the amino acid sequences of the complementarity determining regions (CDRs) are defined by the Kabat coding table and are listed in Table 3. Sequence comparison analysis indicated that the CDRs of anti-HER2 mRC48 were significantly different from the known HER2 antibodies including Herceptin (trastuzumab).
  • VH Heavy chain
  • VL Light chain
  • VL CDR1 DYYIH (SEQ ID NO.1) KASQDVGTAVA (SEQ ID NO. 4)
  • CDR2 RVNPDHGDSYYNQKFKD SEQ ID NO. 2)
  • WASIRHT SEQ ID NO. 5
  • CDR3 ARNYLFDHW SEQ ID NO. 3
  • HQFATYT SEQ ID NO. 6
  • the humanized anti-HER2 monoclonal antibody RC48 was obtained by CDR grafting, and the heavy or light chain variable region was directly synthesized by Nanjing Kingsray Biotechnology Co., Ltd.
  • the synthesized variable region including the Kozak consensus sequence
  • the start codon, the heavy or light chain signal peptide, the human framework region and the murine CDR, the variable region and the human IgG1 k constant region are joined into a complete fragment by overlapping extension PCR.
  • VH1 5'CGCGGATCC GCCGCCACCATGGGATGGAGCT3' (SEQ ID NO: 13)
  • VH2 5'GATGGGCCCTTGGTGCTAGCGGAGCTCACTGTCACCAGTGTT3' (SEQ ID NO: 14)
  • VL1 5'CGCGGATCC GCCGCCACCATGGACATGAGGGT 3' (SEQ ID NO: 17)
  • VL2 5'GATGGTGCAGCCACAGTACGCTTTATCTCAACTTTTG T
  • CL2 5'CCGGAATTCACACTCTCCCCTGTTGAAGC3' (SEQ ID NO: 20)
  • the synthetic variable region was used as a template
  • VH1 and VH2 were used as primers
  • human IgG1 ⁇ heavy chain constant region was used as a template
  • CH1 and CH2 were used as primers to amplify the variable region of heavy chain
  • the constant region and the amplification conditions were: denaturation at 94 ° C for 30 s, annealing at 60 ° C for 30 s, extension at 72 ° C for 1 minute, circulation 30 times, and finally extension at 72 ° C for 10 minutes.
  • the heavy chain sequence of RC48 was amplified by using two PCR products as templates and VH1 and CH2 as primers.
  • the amplification conditions were: denaturation at 94 ° C for 30 s, annealing at 60 ° C for 30 s, extension at 72 ° C for 2 minutes, circulation 30 times, and finally extension at 72 ° C for 10 minutes.
  • variable and constant regions of the light chain were amplified, and the amplification conditions were: denaturation at 94 ° C for 30 s, annealing at 60 ° C for 30 s, extension at 72 ° C for 1 minute, circulation 30 times, and finally extension at 72 ° C for 10 minutes.
  • the light chain sequence was amplified by using two PCR products as templates and VL1 and CL2 as primers.
  • the amplification conditions were: denaturation at 94 ° C for 30 s, annealing at 60 ° C for 30 s, extension at 72 ° C for 2 minutes, circulation 30 times, and finally extension at 72 ° C for 10 minutes.
  • RC48 comprises human IgG1 kappa heavy chain constant region and heavy chain variable region RC48-VH, and human IgG1 kappa light chain constant region and light chain variable region RC48-VL .
  • the human-mouse chimeric antibody cRC48 was also obtained by the same method, and the murine variable region and the human IgG1 k constant region were ligated into a complete fragment by overlap extension PCR.
  • cRC48 The chimeric anti-HER2 RC48 (referred to as cRC48) is composed of a murine-human chimeric cRC48 heavy and light chain. RC48 includes the humanized heavy chain RC48-VH and the humanized light chain RC48-VL.
  • Both cRC48 and RC48 were able to express, and the antibodies were collected from CHO cell supernatants, purified by Protein A, and analyzed by SDS-PAGE under reducing and non-reducing conditions (see Figure 2).
  • CHO cells secreting RC48 antibody as described above ie, CHO cells transfected with human IgG1 kappa heavy chain constant region and heavy chain variable region RC48-VH, and human IgG1 kappa light chain constant region and light chain variable region RC48-VL
  • the HER2-binding affinity constant (Kd) of chimeric cRC48 and humanized RC48 antibody (RC48) was determined by ELISA. The specific method can be seen in Example 1, that is, coating 96-well plates with soluble HER2-ECD protein, followed by dilution. Antibodies (Herceptin and chimeric cRC48 as controls) were incubated with antibodies to HER2-ECD (all forms of human IgG1 kappa) using HRP-conjugated sheep F(ab') 2 anti-human IgG Fc-specific The secondary antibody was tested (invitrogen).
  • the humanized anti-HER2 antibody RC48 has a higher HER2-ECD binding affinity than the cRC48 (mean affinity constant 77 pM) and Herceptin (mean affinity constant 97 pM), mean pro The constant is 44 pM and the results are shown in Table 4.
  • Flow cytometry was used to detect the binding of HER2 endogenously expressed in human breast cancer cells to the humanized anti-HER2 antibody RC48.
  • the results are shown in Fig. 3. 6 ⁇ g of control human IgG, Herceptin, cRC48, RC48 were incubated with two HER2 + cell lines, human breast cancer cells SK-BR-3, BT474, and HER2 - cell MDA-MB468 (2 ⁇ 10 7 cells), respectively. Incubate for 30-45 minutes.
  • the binding curve of the cell-based anti-HER2 antibody to the cell surface HER2 was obtained by titrating the concentration of the anti-HER2 antibody and the number of cells analyzed in flow cytometry. The results are shown in Figure 4b.
  • the humanized anti-HER2 antibody RC48 showed significant binding affinity, and the binding affinity to HER2 on the surface of BT474 cells was 4 nM, Herceptin and cRC48 were 10 nM and 5 nM, respectively.
  • Table 5 The results are shown in Table 5.
  • Herceptin, cRC48, and RC48 were determined by ELISA. See Example 1 for the ELISA method. 96-well plates were coated with antigens EGFR, HER2, HER3, and HER4, respectively, and the amount of each well was 20 ng. The cells were incubated with different anti-HER2 antibodies, namely Herceptin, cRC48, RC48 antibody, and then horseradish peroxidase. The combined sheep F(ab') 2 anti-mouse IgG Fc specific secondary antibody (Invitrogen) was tested. The results are shown in Figure 5. The results showed that Herceptin, cRC48, and RC48 antibodies had almost no binding to EGFR, HER3, and HER4, but had strong binding to HER2, indicating that Herceptin and RC48 have high specificity for HER2 binding.
  • the human breast cancer BT474 nude mice xenograft model was established by inoculating BT474 cells into the skin of nude mice. After 3 consecutive generations in vivo, the tumor tissues in the vigorous growth period were cut into 1.5 mm3, and inoculated under sterile conditions. In the nude mice (provided by Shanghai Slack Laboratory Animal Co., Ltd., certificate number 2007000540582, license number SCXK (Shanghai) 2012-0002), the right armpit was subcutaneous. The nude mice xenografts were measured with a vernier caliper to measure the diameter of the transplanted tumors, and the animals were randomly divided into groups after the tumors were grown to 100-300 mm3. The test drugs huIgG1, Herceptin, and RC48 were administered at 10 mg/kg each, and the second administration was performed one week later, and the third administration was performed two weeks later, and a total of three administrations were administered.
  • the monoclonal antibody of RC48 was captured from CHO cell culture medium by Protein A, and the purity of SDS-PAGE electrophoresis and SEC analysis was over 95%.
  • the obtained antibody protein was ultrafiltered into a PBS buffer by a 30 KD membrane package, concentrated, and calibrated with a UV spectrophotometer for subsequent coupling reaction.
  • the reducing agent and the protective agent were prepared by using PBS buffer as follows: 1-20 mmol/L TCEP (Tris-2-carboxyethyl-phosphine), 1-20 mmol/L DTPA (Diethylene triamine pentacetate acid) mother liquor, and the amount of reducing agent according to the required couple
  • the combination rate can be added within a certain concentration range, and a certain concentration of monoclonal antibody (for example, 5-30 mg/ml) is mixed according to a certain volume ratio (such as 1:1), and the final concentration molar ratio of TCEP to antibody is 0.5-6.0: 1.
  • the reaction was stirred at 25 ° C for 2 h.
  • the free thiol concentration was measured by DTNB method at 412 nm, and the molar ratio to the antibody was calculated to calculate the number of free thiol groups.
  • the TCEP reduction is reproducible, and the number of free thiol groups after reduction can reach 1.0-8.0.
  • the antibody can be directly coupled after TCEP reduction.
  • Prepare a certain concentration (10mM) drug (vc-MMAE, vc-MMAF, mc-MMAF) (purchased from Shanghai Qianyuan Chemical Technology Co., Ltd.) dissolved in 25% DMSO (dimethyl sulfoxide, dimethyl sulfoxide), according to the drug and The thiol group was slowly added at a molar ratio of 0.3 to 2.8:1, and the reaction was stirred at 25 ° C for 2 hours.
  • the free sulfhydryl concentration (close to 0) was detected by DTNB method at 412 nm, and Sephadex G-25 was purified to remove residual unreacted drugs and free small molecules such as DMSO.
  • ELISA plates were coated with recombinant protein HER2-ECD (concentration 0.5 mg/ml), overnight at 2-8 °C. The plate washer washes the plate 3 times. The 3% BSA-PBST solution was blocked at 37 degrees 2 h. The plate washer washes the plate 3 times. Loading: Dilute the line with PBST solution and dilute 11 points from 1000 ng/ml, 100 ⁇ l/well, 37 ° 2 h. The plate washer washes the plate 3 times. The secondary antibody (goat anti-human IgG-Fc-HRP) was diluted 5000 times with PBST solution. Add TMB coloring solution to develop color, and color at room temperature for 8-10 minutes. The assay was terminated with 2M H 2 SO 4 and read at 450/655 nm. The results are shown in Table 6.
  • HER2-positive breast cancer cells SK-BR-3 and HER2-positive ovarian cancer cells SK-OV-3 were respectively treated with DMEM containing 10% fetal bovine serum.
  • the McCoy's 5A medium (the medium was purchased from Gibco) was resuspended, inoculated into a 96-well plate at a density of 5000, 4000/well, and cultured in a 37 ° C, 5% CO 2 incubator for 24 hours.
  • the results are shown in Fig. 12 to Fig. 13. It can be seen from the figure that the inhibitory effect of the ADC drug of the present invention on each cell is significantly stronger than that of the naked antibody drug at the same concentration, and the inhibition rate on cell proliferation can be increased by half.
  • the MMAF-conjugated ADC drug according to the present invention inhibits SK-BR-3, SK-OV-3 cells more strongly than the MMAE-coupled ADC according to the present invention. Drugs, but both were significantly better than the positive control T-DM1.
  • BT474 cells 5 million BT474 cells were suspended in PBS and inoculated into BALB/c nude mice (provided by Shanghai Slack Laboratory Animal Co., Ltd., certificate number 2007000540582, license number SCXK (Shanghai) 2012-0002).
  • the tumor tissue in the vigorous growth period was cut into 1.5 mm 3 and inoculated subcutaneously in the right axilla of the nude mice under aseptic conditions.
  • the nude mice xenografts were measured with a vernier caliper to measure the diameter of the transplanted tumors, and the animals were randomly divided into groups after the tumors were grown to 100-300 mm3.
  • the tested drugs were administered with RC48 10 mg/kg, RC48-vc-MMAE 10 mg/kg, RC48-vc-MMAF 10 mg/kg, RC48-mc-MMAF 10 mg/kg for 3 times; the negative control group was given the same amount of physiology. brine.
  • the tumor volume of the negative control group reached 485 mm 3 at 37 days after administration, and the tumor volume of the RC48 group was 83% of the control group, indicating that RC48 had a certain inhibitory effect on BT474 tumor growth.
  • the three antibody conjugates tested in this experiment, RC48-vc-MMAE, RC48-vc-MMAF, and RC48-mc-MMAF all significantly inhibited the growth of BT474 tumors, and the tumor volume was reduced to the control group at 37 days after administration. -19%. During the 37-day experiment, 3 mice died in the control group, while the RC48-vc-MMAE group survived.
  • BT474/L1.9 is a BT-474 cell Long-term treatment with lapatinib And lapatinib resistance.
  • SPF-class BALB/c nude mice were subcutaneously inoculated with a certain number of BT-474/L1.9 tumor cells. After the tumors were grown to 100-200 mm3, the animals were randomly divided into groups. The doses administered are shown in the figure, RC48-vc-MMAE, Kadcyla TM (purchased from Roche Pharmaceuticals), once a week, a total of 2 times, (purchased from Roche Pharmaceuticals) was administered once a week for 3 times, and lapatinib (purchased from GSK) was administered daily. The tumor volume was measured twice a week, and the mice were weighed to observe the drug tolerance of the tumor-bearing mice, and the data were recorded. The tumor volume of each tumor-bearing mouse at different observation time points and the tumor inhibition rate of each tumor-bearing mouse were calculated.
  • Figure 15 shown in (10mg/kg) inhibited tumor rate of subcutaneous xenografts of BT474/L1.9 nude mice by 51%; lapatinib (200mg/kg) inhibited tumor rate of BT474/L1.9 by 45%; Cancer BT474/L1.9 pair And lapatinib are resistant.
  • RC48-vc-MMAE (1.5, 5 mg/kg) dose-dependently inhibited the growth of subcutaneous xenografts in BT474/L1.9 nude mice, with tumor inhibition rates of 38% and 91%, respectively; and reference drug Kadcyla TM (5 mg/kg ) kg) subcutaneously BT474 / L1.9 nude mice, the tumor inhibition rate was 58%, indicating BT474 / L1.9 also Kadcyla TM resistant. Tumor-bearing mice are well tolerated by the above drugs.
  • the ADC of the present invention showed significant antitumor activity against the subcutaneous xenograft model of BT474/L1.9 cells in nude mice; significantly stronger than Herceptin and lapatinib (P ⁇ 0.01); at the same dose compared with Kadcyla TM, antitumor effect RC48-vc-MMAE present invention has obvious advantages (P ⁇ 0.01) at 5mg / kg, the inhibition rate of 91%: 58%.
  • Human ovarian cancer SK-OV-3 xenograft model was obtained by inoculating SK-OV-3 cells into nude mice (Shanghai Slack Laboratory Animal Co., Ltd., certificate number 2007000540582, license number SCXK (Shanghai) 2012- 0002) Established under the skin. Animals were randomized after tumor growth to 100-300 mm3 .
  • the tested drugs were administered RC4810mg/kg, T-DM110mg/kg, RC48-vc-MMAE 3mg/kg, RC48-vc-MMAE 10mg/kg, RC48-vc-MMAF 3mg/kg, RC48-vc-MMAF10mg/kg, RC48 -mc-MMAF 3 mg/kg, RC48-mc-MMAF 10 mg/kg, once a week, a total of three times; paclitaxel 10 mg/kg, three times a week for three weeks; the negative control was given the same amount of normal saline.
  • the anti-tumor effect of RC48 on SK-OV-3 after binding to different linkers and therapeutic agents showed that the ADC of the present invention had a good anti-tumor effect compared to the unconjugated naked antibody RC48, at the same dose of 10 mg/
  • the anti-tumor effect of the RC48 conjugate of the present invention has a significant advantage over T-DM1 at kg. The specific results are shown in Figure 16.
  • Reference drug (10mg/kg, 3 times a week) can inhibit the growth of subcutaneous xenografts in NCI-N87 nude mice, but the tumor inhibition rate is only 49%; another reference drug small molecule EGFR/HER2 inhibitor Lapa Tinidil (200 mg/kg, once a day for 21 days) was effective against subcutaneous xenografts of NCI-N87 nude mice with a tumor inhibition rate of 78%. Tumor-bearing mice are well tolerated by the above drugs, and no symptoms such as weight loss occur.
  • Quarantine-qualified KM mice were randomly divided into the following dose groups: MMAE 2 mg/kg, MMAF 50 mg/kg, RC48-vc-MMAE 25 mg/kg (corresponding MMAE dose 0.5 mg/kg) to 200 mg ( MMAE dose is 4.0mg/kg), RC48-vc-MMAF 50mg/kg (MMAF dose is 1.0mg/kg) ⁇ 450mg/kg (MMAF dose is 9.0mg/kg), RC48-mc-MMAF50mg/kg (MMAF dose) There are 13 groups of different doses of 1.0mg/kg) to 450mg/kg (MMAF dose of 9.0mg/kg), male and female, and the corresponding drug solution is injected intravenously.
  • mice Another batch of mice was used as a control group, and 0.9% sodium chloride injection was administered in the same volume of tail vein. Weigh once every other day after administration and continue until the 16th day.
  • the weight gain curves of the MMAE or MMAF series mice are shown in Figure 18, respectively.
  • the test results showed that under the conditions of this experiment, the toxicity of RC48-vc-MMAE was less than that of unconjugated MMAE at the same dose.
  • the maximum tolerated dose of RC48-vc-MMAE mice 100 mg/kg (MMAE dose 2.0 mg/kg) to 150 mg/kg (MMAE dose 3.0 mg/kg).
  • the RC48-vc-MMAF toxicity was significantly greater than that of RC48-mc-MMAF at the same dose, and the maximum tolerated dose of RC48-vc-MMAF was 50 mg/kg (MMAF dose 1.0 mg/kg) to 100 mg/kg (MMAF). The dose is between 2.0 mg/kg).
  • the mouse MTD of RC48-mc-MMAF was between 150 mg/kg (MMAF dose 3.0 mg/kg) to 450 mg/kg (MMAF dose 9.0 mg/kg).
  • SPF grade SD rats (certified certificate number SCXK (Beijing) 2012-0001) purchased from Beijing Weitong Lihua Experimental Animal Technology Co., Ltd. were randomly divided into 7 groups according to gender segments, 10 in each group, half male and half female; The 7 groups were given saline negative control group, 0.48 mg/kg MMAE control group, 40 mg/kg RC48 naked anti-control group and series RC48-vc-MMAE dose group. The rats were administered by tail vein infusion. The period was 21 days; the D1 (pre-drug), D8, D15 and D22 were weighed. After the observation period, the animals were euthanized, gross pathological observations were performed, and the abnormal tissues and organs were examined for histological examination. .
  • MMAE control group 9/10 animals, RC48-vc-MMAE 30 mg/kg group 1/10 animals and 40 mg/kg group 2/10 animals were found dead during the test period, and no death or sputum was observed in the other groups. Dead state.
  • the lethal dose of RC48-vc-MMAE rats was 30 mg/kg, and the maximum tolerated dose was greater than or equal to 24 mg/kg (approximately conjugated MMAE dose was 0.48 mg/kg).
  • SD rats were given a single intravenous infusion of RC48 naked antibody at a dose of 40 mg/kg. No obvious toxicity symptoms were observed.
  • the maximum tolerated dose of the rats was greater than or equal to 40 mg/kg.
  • T-DM1 trastuzumab emtansine

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Organic Chemistry (AREA)
  • Epidemiology (AREA)
  • Genetics & Genomics (AREA)
  • Cell Biology (AREA)
  • Oncology (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Microbiology (AREA)
  • Biomedical Technology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Mycology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
PCT/CN2014/091332 2013-11-19 2014-11-18 抗her2抗体及其缀合物 WO2015074528A1 (zh)

Priority Applications (16)

Application Number Priority Date Filing Date Title
AU2014352475A AU2014352475B2 (en) 2013-11-19 2014-11-18 Anti-HER2 antibody and conjugate thereof
EP18204950.2A EP3480215B8 (en) 2013-11-19 2014-11-18 Anti-her2 antibody and conjugate thereof
DK14864053.5T DK3072907T3 (en) 2013-11-19 2014-11-18 ANTI-HER2- ANTIBODY AND CONJUGATE THEREOF
US15/037,104 US10087260B2 (en) 2013-11-19 2014-11-18 Anti-HER2 antibody and conjugate thereof
CA2919359A CA2919359C (en) 2013-11-19 2014-11-18 Anti-her2 antibody and conjugate thereof
JP2016537121A JP6326137B2 (ja) 2013-11-19 2014-11-18 抗her2抗体及びその結合体
CN201811053363.6A CN110240655B (zh) 2013-11-19 2014-11-18 抗her2抗体及其缀合物
KR1020167012018A KR101854443B1 (ko) 2013-11-19 2014-11-18 항-her2 항체 및 이의 접합체
BR112016002752-3A BR112016002752B1 (pt) 2013-11-19 2014-11-18 Anticorpo ou seus fragmentos funcionais que pode especificamente se ligar ao her2, conjugados, composição farmacêutica, e, uso do anticorpo ou seus fragmentos funcionais, do conjugado ou da composição farmacêutica
RU2016106339A RU2656161C1 (ru) 2013-11-19 2014-11-18 Анти-неr2 антитело и его конъюгат
EP14864053.5A EP3072907B1 (en) 2013-11-19 2014-11-18 Anti-her2 antibody and conjugate thereof
PL18204950T PL3480215T3 (pl) 2013-11-19 2014-11-18 Przeciwciało anty-HER2 i jego koniugat
CN201480006648.8A CN105008398B (zh) 2013-11-19 2014-11-18 抗her2抗体及其缀合物
KR1020187012035A KR101936697B1 (ko) 2013-11-19 2014-11-18 항-her2 항체 및 이의 접합체
KR1020187036881A KR101993136B1 (ko) 2013-11-19 2014-11-18 항-her2 항체 및 이의 접합체
CY20211100704T CY1124651T1 (el) 2013-11-19 2021-08-06 Αντισωμα εναντι toy her2 και συζευγμα αυτου

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201310586326 2013-11-19
CN201310586326.2 2013-11-19

Publications (1)

Publication Number Publication Date
WO2015074528A1 true WO2015074528A1 (zh) 2015-05-28

Family

ID=53178934

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2014/091332 WO2015074528A1 (zh) 2013-11-19 2014-11-18 抗her2抗体及其缀合物

Country Status (19)

Country Link
US (1) US10087260B2 (es)
EP (2) EP3072907B1 (es)
JP (2) JP6326137B2 (es)
KR (3) KR101854443B1 (es)
CN (3) CN109320612B (es)
AU (1) AU2014352475B2 (es)
CA (1) CA2919359C (es)
CY (1) CY1124651T1 (es)
DK (2) DK3072907T3 (es)
ES (1) ES2879799T3 (es)
HR (1) HRP20211023T1 (es)
HU (1) HUE055269T2 (es)
LT (1) LT3480215T (es)
PL (1) PL3480215T3 (es)
PT (1) PT3480215T (es)
RS (1) RS62157B1 (es)
RU (1) RU2656161C1 (es)
SI (1) SI3480215T1 (es)
WO (1) WO2015074528A1 (es)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017179862A1 (ko) * 2016-04-12 2017-10-19 앱클론(주) 안정성이 개선된 her2에 특이적으로 결합하는 항체
WO2020042941A1 (zh) * 2018-08-29 2020-03-05 荣昌生物制药烟台有限公司 抗her2抗体药物偶联物在治疗尿路上皮癌中的用途
WO2020177570A1 (zh) 2019-03-01 2020-09-10 荣昌生物制药(烟台)股份有限公司 一种Her2伴随诊断免疫组化检测抗体及其应用
WO2020192693A1 (zh) 2019-03-26 2020-10-01 荣昌生物制药(烟台)股份有限公司 抗Her2抗体药物偶联物药物制剂
WO2020233534A1 (zh) * 2019-05-17 2020-11-26 百奥泰生物制药股份有限公司 抗体-药物偶联物制剂、制备方法及应用
WO2022174775A1 (en) * 2021-02-18 2022-08-25 Remegen Co., Ltd. Use of antibody-drug conjugate targeting her2 in treatment of specific breast cancer

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PL3480215T3 (pl) * 2013-11-19 2021-12-13 Remegen Co., Ltd. Przeciwciało anty-HER2 i jego koniugat
BR112021005320A2 (pt) 2018-12-17 2021-06-15 Remegen Co., Ltd. ligante para conjugados de anticorpo-fármaco e seu uso
CN109498816A (zh) * 2019-01-07 2019-03-22 合肥瀚科迈博生物技术有限公司 一种抗体偶联药物的制备方法
KR20220115566A (ko) 2019-11-15 2022-08-17 시아겐 인크. 항-her2 항체-약물 접합체와 조합된 투카티닙으로 her2 양성 유방암을 치료하는 방법
MX2022009138A (es) * 2020-01-23 2022-10-27 Bioatla Inc Anticuerpos y fragmentos de anticuerpos anti-her2 condicionalmente activos, sus inmunoconjugados y usos de estos.
CN112402616A (zh) * 2020-11-23 2021-02-26 荣昌生物制药(烟台)股份有限公司 一种含环丙沙星和抗体药物偶联物的药物组合物及其应用
CN112353947A (zh) * 2020-11-23 2021-02-12 荣昌生物制药(烟台)股份有限公司 一种含二甲双胍和抗体药物偶联物的药物组合物及其应用
CN114796519A (zh) * 2021-01-27 2022-07-29 南开大学 一种蛋白质偶联小分子药物的制备和应用
US11814394B2 (en) * 2021-11-16 2023-11-14 Genequantum Healthcare (Suzhou) Co., Ltd. Exatecan derivatives, linker-payloads, and conjugates and thereof
CN114736300B (zh) * 2022-06-09 2022-08-19 苏州百道医疗科技有限公司 一种抗her2重组兔单克隆抗体及其应用

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1993146A (zh) 2004-06-01 2007-07-04 健泰科生物技术公司 抗体-药物偶联物和方法
CN101143902A (zh) * 2007-05-18 2008-03-19 中国医学科学院医药生物技术研究所 抗HER2单链抗体-力达霉素强化融合蛋白HER2(Fv-LDM)
CN100482281C (zh) 1999-06-25 2009-04-29 基因技术股份有限公司 抗ErbB抗体-类美坦素偶联物在制备药物中的应用
CN102027135A (zh) * 2008-03-14 2011-04-20 健泰科生物技术公司 与药物抗性有关的遗传变异

Family Cites Families (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU784045B2 (en) * 1999-06-25 2006-01-19 Genentech Inc. Humanized anti-ErbB2 antibodies and treatment with anti-ErbB2 antibodies
US20040132101A1 (en) * 2002-09-27 2004-07-08 Xencor Optimized Fc variants and methods for their generation
KR101200133B1 (ko) * 2004-06-01 2012-11-13 제넨테크, 인크. 항체 약물 접합체 및 방법
KR20070038557A (ko) 2004-07-22 2007-04-10 제넨테크, 인크. Her2 항체 조성물
AU2008251608B2 (en) 2007-05-08 2014-03-27 Genentech, Inc. Cysteine engineered anti-MUC16 antibodies and antibody drug conjugates
WO2008150485A2 (en) * 2007-05-29 2008-12-11 Wyeth Erbb2 binding proteins and use thereof
UA127472C2 (uk) * 2008-03-18 2023-09-06 Дженентек, Інк. Комбінація кон'югата анти-her2-антитіло-лікарський засіб і хіміотерапевтичного засобу і спосіб застосування
US8750701B2 (en) * 2008-09-19 2014-06-10 Telefonaktiebolaget L M Ericsson (Publ) Protection mechanism for a communications network
CN102030827B (zh) 2009-09-25 2014-09-24 上海抗体药物国家工程研究中心有限公司 一种高亲和力的抗her2单克隆抗体
US8937159B2 (en) * 2009-12-16 2015-01-20 Abbvie Biotherapeutics Inc. Anti-HER2 antibodies and their uses
CN102167742B (zh) * 2010-02-25 2014-05-14 上海百迈博制药有限公司 一种全人源抗her2单克隆抗体、其制备方法及用途
TW201141519A (en) * 2010-03-04 2011-12-01 Symphogen As Anti-HER2 antibodies and compositions
US8609095B2 (en) 2010-03-04 2013-12-17 Symphogen A/S Anti-HER2 antibodies and compositions
CA3051311A1 (en) 2010-05-27 2011-12-01 Genmab A/S Monoclonal antibodies against her2
CN103153339B (zh) 2010-05-27 2021-05-04 根马布股份公司 针对her2表位的单克隆抗体
US9193791B2 (en) * 2010-08-03 2015-11-24 City Of Hope Development of masked therapeutic antibodies to limit off-target effects
AU2013255537B2 (en) 2012-05-02 2018-02-15 Symphogen A/S Humanized pan-her antibody compositions
PL3480215T3 (pl) * 2013-11-19 2021-12-13 Remegen Co., Ltd. Przeciwciało anty-HER2 i jego koniugat

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN100482281C (zh) 1999-06-25 2009-04-29 基因技术股份有限公司 抗ErbB抗体-类美坦素偶联物在制备药物中的应用
CN1993146A (zh) 2004-06-01 2007-07-04 健泰科生物技术公司 抗体-药物偶联物和方法
CN101143902A (zh) * 2007-05-18 2008-03-19 中国医学科学院医药生物技术研究所 抗HER2单链抗体-力达霉素强化融合蛋白HER2(Fv-LDM)
CN102027135A (zh) * 2008-03-14 2011-04-20 健泰科生物技术公司 与药物抗性有关的遗传变异

Non-Patent Citations (27)

* Cited by examiner, † Cited by third party
Title
BASELGA J.: "Annals of Oncology", vol. 21, 2010, article "Treatment of HER2-overexpressing breast cancer", pages: VII36 - VII40
BEBBINGTON ET AL., BIO/TECHNOLOGY, vol. 10, 1992, pages 169 - 175
CHEN, Y.Z. ET AL.: "Expressions and Significance of HER2/neu protein in Epithelial Ovarian Carcinoma and its Metastatic lesions", CHINESE GENERAL PRACTICE, vol. 12, 2009, pages 1268 - 1271
DE BONO JS; BELLMUNT J; ATTARD G; DROZ JP; MILLER K; FLECHON A; STERNBERG C; PARKER C; ZUGMAIER G; HERSBERGER-GIMENEZ V: "Open-label phase II study evaluating the efficacy and safety of two doses of pertuzumab in castrate chemotherapy-naive patients with hormone-refractory prostate cancer", J CLIN ONCO, vol. 25, no. 3, 20 January 2007 (2007-01-20), pages 257 - 262
DE VRIES JF; ZWAAN CM; DE BIE M; VOERMAN JS; DEN BOER ML; VAN DONGEN JJ; VAN DER VELDEN VH.: "The novel calicheamicin-conjugated CD22 antibody inotuzumab ozogamicin (CMC-544) effectively kills primary pediatric acute lymphoblastic leukemia cells", LEUKEMIA, vol. 26, no. 2, February 2012 (2012-02-01), pages 255 - 264
DOSIO F P. BRUSA; L CATTEL.: "Immunotoxins and anticancer drugs conjugate assemblies: the role of the linkage between components", TOXINS, vol. 3, 2011, pages 848 - 883, XP055107282, DOI: doi:10.3390/toxins3070848
GIRISH S; GUPTA M; WANG B ET AL.: "Clinical pharmacology of trastuzumab emtansine (T-DM1): an antibody-drug conjugate in development for the treatment of HER2-positive cancer", CANCER CHEMOTHERAPY AND PHARMACOLOGY, vol. 69, 2012, pages 1229 - 1240, XP035047349, DOI: doi:10.1007/s00280-011-1817-3
HARLOW; LANE: "Antibodies: A Laboratory Manual", 1988, COLD SPRING HARBOR LABORATORY, pages: 726
HUANG, X.E.: "The Study on the expression of HER2 and its correlation with the prognosis of breast carcinoma", CLINICAL RESEARCH, vol. 49, 2008, pages 39 - 40
JONES ET AL., NATURE, vol. 321, 1986, pages 522 - 525
JOSEPH A. FRANCISCO; CHARLES G. CERVENY; DAMON L. MEYER; BRUCE J. MIXAN; KERRY KLUSSMAN; DANA, F. CHACE; STARR X. REJNIAK; KRISTIN: "cACIO-vcM-MAE, an anti-CD30-monomethyl auristatin E conjugate with Potent and selective antitumor activity", BLOOD, vol. 102, 2003, pages 1458 - 1465
JOURNAL OF IMMUNOLOGICAL METHODS, vol. 270, 2002, pages 155 - 162
KABAT ET AL.: "Sequences of proteins of immunological interest", 1991, NIH
KATZ J; JANIK JE; YOUNES A.: "2011 Brentuximab Vedotin (SGN-35).", CLIN CANCER RES, vol. 17, no. 20, 15 October 2011 (2011-10-15), pages 6428 - 6436
KOHLER; MILSTEIN, NATURE, vol. 256, 1975, pages 495 - 497
KROP IE; BEERAM M; MODI S ET AL.: "Phase I Study of Trastuzumab-DM1, an HER2 Antibody-Drug Conjugate, Given Every 3 Weeks to Patients With HER2-Positive Metastatic Breast Cancer", JOURNAL OF CLINICAL ONCOLOGY, vol. 28, 2010, pages 2698 - 2704, XP055156437, DOI: doi:10.1200/JCO.2009.26.2071
LEWIS PHILLIPS GD; LI G; DUGGER DL ET AL.: "Targeting HER2-Positive Breast Cancer with Trastuzumab-DMI, an Antibody-Cytotoxic Drug Conjugate", CANCER RESEARCH, vol. 68, 2008, pages 9280 - 9290, XP055013498, DOI: doi:10.1158/0008-5472.CAN-08-1776
MOUNTAIN ET AL., BIOTECHNOL. GENET. ENG. REV., vol. 10, 1992, pages 1 - 142
NAHTA, R.; F.J.ESTEVA: "Trastuzumab triumphs and tribulations", ONCOGENE, vol. 6, no. 25, 2007, pages 3637 - 3643, XP002458380, DOI: doi:10.1038/sj.onc.1210379
NEDDLEMAN; WUNSCH, J. MOL. BIOL., vol. 48, 1970, pages 443
PEARSON; LIPMAN, PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 2444
PETERSDORF SH; KOPECKY KJ; SLOVAK M; WILLMAN C; NEVILL T; BRANDWEIN J; LARSON RA; ERBA HP; STIFF PJ; STUART RK: "A phase III study of gemtuzumab ozogamicin during induction and post-consolidation therapy in younger patients with acute myeloid leukemia", BLOOD, 16 April 2013 (2013-04-16)
RIECHMANN ET AL., NATURE, vol. 332, 1988, pages 323 - 327
SINGER ET AL., J. IMMUN, vol. 150, 1992, pages 2844 - 2857
SMITH; WATERMAN, AD. APP. MATH., vol. 2, 1981, pages 482
VERHOEYEN ET AL., SCIENCE, vol. 239, 1988, pages 1534 - 1536
VERHOEYN ET AL., BIOESSAYS, vol. 8, 1988, pages 74

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017179862A1 (ko) * 2016-04-12 2017-10-19 앱클론(주) 안정성이 개선된 her2에 특이적으로 결합하는 항체
US11248059B2 (en) 2016-04-12 2022-02-15 Abclon Inc. Antibody having improved stability and specifically binding to HER2
WO2020042941A1 (zh) * 2018-08-29 2020-03-05 荣昌生物制药烟台有限公司 抗her2抗体药物偶联物在治疗尿路上皮癌中的用途
TWI767139B (zh) * 2018-08-29 2022-06-11 中國大陸商榮昌生物製藥(煙臺)股份有限公司 抗her2抗體藥物偶聯物在治療尿路上皮癌中的用途
WO2020177570A1 (zh) 2019-03-01 2020-09-10 荣昌生物制药(烟台)股份有限公司 一种Her2伴随诊断免疫组化检测抗体及其应用
WO2020192693A1 (zh) 2019-03-26 2020-10-01 荣昌生物制药(烟台)股份有限公司 抗Her2抗体药物偶联物药物制剂
CN113365665A (zh) * 2019-03-26 2021-09-07 荣昌生物制药(烟台)股份有限公司 抗Her2抗体药物偶联物药物制剂
TWI825297B (zh) * 2019-03-26 2023-12-11 中國大陸商榮昌生物製藥(煙臺)股份有限公司 抗Her2 抗體藥物偶聯物藥物製劑
EP4316521A2 (en) 2019-03-26 2024-02-07 RemeGen Co., Ltd. Anti-her2 antibody drug conjugate pharmaceutical preparation
WO2020233534A1 (zh) * 2019-05-17 2020-11-26 百奥泰生物制药股份有限公司 抗体-药物偶联物制剂、制备方法及应用
WO2022174775A1 (en) * 2021-02-18 2022-08-25 Remegen Co., Ltd. Use of antibody-drug conjugate targeting her2 in treatment of specific breast cancer

Also Published As

Publication number Publication date
BR112016002752A8 (pt) 2023-02-28
JP2016528902A (ja) 2016-09-23
CN110240655A (zh) 2019-09-17
AU2014352475B2 (en) 2017-08-17
US20160304621A1 (en) 2016-10-20
RS62157B1 (sr) 2021-08-31
CN109320612A (zh) 2019-02-12
LT3480215T (lt) 2021-08-25
JP6728264B2 (ja) 2020-07-22
CA2919359A1 (en) 2015-05-28
DK3480215T3 (da) 2021-07-05
BR112016002752A2 (pt) 2017-11-21
PT3480215T (pt) 2021-07-05
KR20190002716A (ko) 2019-01-08
EP3072907A4 (en) 2017-06-14
HUE055269T2 (hu) 2021-11-29
US10087260B2 (en) 2018-10-02
JP2018138034A (ja) 2018-09-06
RU2656161C1 (ru) 2018-05-31
CY1124651T1 (el) 2022-07-22
DK3480215T5 (da) 2021-08-16
EP3072907B1 (en) 2019-02-20
JP6326137B2 (ja) 2018-05-16
EP3480215B1 (en) 2021-06-23
CA2919359C (en) 2019-01-15
CN110240655B (zh) 2023-05-16
KR101993136B1 (ko) 2019-06-26
EP3480215A1 (en) 2019-05-08
EP3072907A1 (en) 2016-09-28
KR20160065972A (ko) 2016-06-09
CN105008398A (zh) 2015-10-28
ES2879799T3 (es) 2021-11-23
PL3480215T3 (pl) 2021-12-13
AU2014352475A1 (en) 2016-02-18
CN109320612B (zh) 2020-10-27
HRP20211023T1 (hr) 2021-09-17
SI3480215T1 (sl) 2021-12-31
RU2016106339A (ru) 2017-12-25
KR20180050423A (ko) 2018-05-14
CN105008398B (zh) 2018-10-16
EP3480215B8 (en) 2021-07-28
KR101854443B1 (ko) 2018-06-15
DK3072907T3 (en) 2019-04-01
KR101936697B1 (ko) 2019-01-10

Similar Documents

Publication Publication Date Title
JP6728264B2 (ja) 抗her2抗体及びその結合体
KR20200120728A (ko) 항-메소텔린 항체 및 이의 항체 약물 콘쥬게이트
TWI767139B (zh) 抗her2抗體藥物偶聯物在治療尿路上皮癌中的用途
CN112175082B (zh) 抗叶酸受体α抗体、其缀合物及其用途
WO2023025243A1 (zh) 抗Nectin-4抗体、药物缀合物及其制备方法和用途
TWI843105B (zh) 抗her2 抗體藥物偶聯物在治療尿路上皮癌中的用途
JP2024086799A (ja) 尿路上皮がんの治療における抗her2抗体-薬物コンジュゲートの使用
BR112016002752B1 (pt) Anticorpo ou seus fragmentos funcionais que pode especificamente se ligar ao her2, conjugados, composição farmacêutica, e, uso do anticorpo ou seus fragmentos funcionais, do conjugado ou da composição farmacêutica

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14864053

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2919359

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2014352475

Country of ref document: AU

Date of ref document: 20141118

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112016002752

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 2016537121

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20167012018

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 15037104

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

REEP Request for entry into the european phase

Ref document number: 2014864053

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2014864053

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2016106339

Country of ref document: RU

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112016002752

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20160210